Clec16a, Nrdp1, and USP8 Form a Ubiquitin-Dependent

Total Page:16

File Type:pdf, Size:1020Kb

Clec16a, Nrdp1, and USP8 Form a Ubiquitin-Dependent Page 1 of 46 Diabetes Clec16a, Nrdp1, and USP8 form a ubiquitin-dependent tripartite complex that regulates beta cell mitophagy Gemma Pearson1; Biaoxin Chai1; Tracy Vozheiko1; Xueying Liu1; Malathi Kandarpa2; 3 1,4* Robert C. Piper ; and Scott A. Soleimanpour 1Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA. 2Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48105, USA. 3Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA 4Veterans Affairs Ann Arbor Health Care System, Ann Arbor, MI 48105, USA. *Corresponding Author Scott A. Soleimanpour, M.D., 1000 Wall Street, 5317 Brehm Tower, Ann Arbor, MI 48105. Phone: (734) 763-0528 Fax: (734) 232-8162 E-mail: [email protected] Running Title: β-cell mitophagy requires ubiquitin signaling Word Count: 4448 Figures: 7 Diabetes Publish Ahead of Print, published online November 27, 2017 Diabetes Page 2 of 46 ABSTRACT Mitophagy is a cellular quality control pathway, which is essential to eliminate unhealthy mitochondria. While mitophagy is critical to pancreatic β-cell function, the post-translational signals governing β-cell mitochondrial turnover are unknown. Here we report that ubiquitination is essential for the assembly of a mitophagy regulatory complex, comprised of the E3 ligase Nrdp1, the deubiquitinase enzyme USP8, and Clec16a, a mediator of β-cell mitophagy with unclear function. We discover that the diabetes gene Clec16a encodes an E3 ligase, which promotes non-degradative ubiquitin conjugates to direct its mitophagy effectors and stabilize the Clec16a-Nrdp1-USP8 complex. Inhibition of the Clec16a pathway by the chemotherapeutic lenalidomide, a selective ubiquitin ligase inhibitor associated with new onset diabetes, impairs β-cell mitophagy, oxygen consumption, and insulin secretion. Indeed, patients treated with lenalidomide develop compromised β-cell function. Moreover, the β-cell Clec16a-Nrdp1-USP8 mitophagy complex is destabilized and dysfunctional following lenalidomide treatment as well as following glucolipotoxic stress. Thus, the Clec16a-Nrdp1- USP8 complex relies on ubiquitin signals to promote mitophagy and maintain mitochondrial quality control necessary for optimal β-cell function. 2 Page 3 of 46 Diabetes Pancreatic β-cells rely on mitochondrial bioenergetics to fuel glucose-stimulated insulin secretion (GSIS), which is essential for normal glucose homeostasis and is impaired in all forms of diabetes (1). Beyond respiratory function, the mass of healthy mitochondria, regulated by a balance of mitochondrial biogenesis and turnover, is vital to maintain energy demands for proper stimulus-secretion coupling. Mitochondrial autophagy (mitophagy) is a quality control mechanism necessary for the selective elimination of dysfunctional mitochondria to maintain optimal mitochondrial respiration in many cell types, including β- cells (2; 3). Despite the importance of mitophagy to β-cell function (4-7), the molecular signals dictating mitophagy and their significance to mitochondrial health remain largely undefined. Ubiquitination of protein substrates, catalyzed by the linkage of activated ubiquitin by three distinct enzymes (E1, E2, E3), leads to diverse responses, including substrate degradation by the ubiquitin-proteasome system, protein trafficking, complex assembly, and cellular signaling (8). Post-translational modification of downstream mitophagy effectors, including the E3 ligase Parkin, by ubiquitination is necessary for efficient mitochondrial turnover (9); however, evidence of post-translational control of proximal mitophagy regulators has not been identified. The E3 ubiquitin ligase neuregulin receptor degradation protein-1 (Nrdp1) and the deubiquitination enzyme ubiquitin specific protease-8 (USP8) counterbalance ubiquitin dynamics to direct Parkin action or its proteasomal degradation (10; 11). Nrdp1 controls Parkin-mediated mitophagy in β-cells through its interaction with, and regulation by, the diabetes gene C-type lectin domain family 16, member A (Clec16a) (4; 5); however, the precise action of Clec16a in mitophagy remains poorly understood. While these factors (Nrdp1, USP8, Clec16a) act upstream as sentinels to ensure mitophagy is utilized appropriately, the role of ubiquitination as a control mechanism for their actions is unknown. 3 Diabetes Page 4 of 46 Here we discover that formation of a ubiquitin-dependent Clec16a-Nrdp1-USP8 complex is necessary for control of mitophagy in β-cells. We identify that inhibition of Clec16a-mediated Nrdp1 ubiquitination by the chemotherapeutic agent lenalidomide impairs Clec16a-mediated mitophagy and, ultimately, reduces β-cell function. We demonstrate that Clec16a is an E3 ubiquitin ligase, which ubiquitinates Nrdp1 to promote assembly of the Clec16a-Nrdp1-USP8 mitophagy complex. Lastly, we determine that mobilization and function of the β-cell Clec16a-Nrdp1-USP8 mitophagy complex is disrupted by glucolipotoxicity. 4 Page 5 of 46 Diabetes RESEARCH DESIGN AND METHODS Patient samples- Samples were provided from de-identified non-diabetic multiple myeloma patients by the University of Michigan Hematologic Malignancies Tissue Bank and in compliance with the University of Michigan IRB. Fasting samples were collected prior to chemotherapy and again 12 weeks later. Patients received a combination of Perifosine, bortezomib, liposomal doxorubicin, Bexxar and/or carfilzomib in the presence/absence of lenalidomide (25 mg PO daily for 28 days x 4 cycles). All patients received corticosteroids (dexamethasone 40 mg PO daily x 12 doses/cycle). C-peptide and glucose concentrations were measured by chemiluminescence and the glucose oxidase method, respectively, by the University of Michigan DRC Clinical Core. Homeostatic model assessment of β-cell function and insulin resistance were calculated by HOMA2 (12). Animals- Animal studies were approved by the University of Michigan IACUC. Clec16aloxP mice (5) were mated to MIP-CreERT2 mice (13) to generate experimental groups. Recombination was induced with 3 IP injections of 150µg/g BW tamoxifen (Cayman Chemical) every other day over 5 days. 10-week old C57BL/6 mice were injected daily with 50mg/kg of lenalidomide IP or vehicle control for 3-weeks. Mice were left to recover for one week before undergoing glucose tolerance testing. Animals were housed on a standard 12-h light/12-h dark cycle with ad libitum access to food and water unless fasted prior to testing. Antibodies- A full listing of antibodies is provided in Supplementary Table 1. Proximity ligation- Studies were performed with a commercially available kit (Duolink; Sigma) per manufacturers’ protocols utilizing Nrdp1 (Bethyl Labs) and USP8 (Sigma) specific antisera. Counter staining to identify β-cells was performed in dissociated human 5 Diabetes Page 6 of 46 islets utilizing PDX-1 antisera (Santa Cruz). Proximity ligation events were quantified via ImageJ similar to published approaches (14). Imaging was performed with an IX81 microscope (Olympus). Z-stack images were captured using an ORCA Flash4 CMOS digital camera (Hamamatsu) and then subjected to deconvolution (CellSens; Olympus). Co- localization analysis was performed as previously described (5). Islet isolation and tissue culture - Primary dermal fibroblasts from WT and Ubc- CreERT;Clec16alox/lox mice were treated ex vivo with tamoxifen as previously described (5). Stably expressing Flag-Clec16a NIH3T3 cells, and Min6 cells (a gift from Dr. Doris Stoffers) were maintained as previously described (5). Mouse islets were isolated and cultured as previously described (15). Cell treatments included dimethylsulfoxide (DMSO; Fisher), lenalidomide (Ark Pharm), chloroquine (Santa Cruz Biotechnology), FCCP (Sigma), bafilomycin A1, MG132, and cycloheximide (all from EMD Millipore). Palmitate/BSA couplings were generated as previously described (16). Human islets– Studies performed on human islets procured from the Integrated Islet Distribution Program (IIDP) were approved by the University of Michigan IRB. Donor information is provided in Supplementary Table 2. Human islets were cultured in PIM(S) (Prodo Laboratories) supplemented with 10% FBS, 1% antibiotic/antimycotic (Life Technologies), and 1% PIM(G) (Prodo Laboratories). Following culture and treatments, islets were dissociated into single cells using trypsin, cytocentrifuged to glass slides, and prepared for immunostaining. Transfections- Transfection of HEK293T, HeLa, and NIH3T3 cells (Lipofectamine 2000; Life Technologies) and Min6 cells (Amaxa Nucleofector) was performed as previously 6 Page 7 of 46 Diabetes described (5). A detailed list of plasmids utilized or generated for this study is found in online supplementary material. Protein expression/purification- Recombinant Clec16a and Nrdp1 was expressed in NiCo21 (DE3) strain E.coli (NEB) following culture in auto-induction media as described (17). GST-tagged and 6xHis-tagged proteins were purified utilizing glutathione matrix or nickel-charged resin (HiCap and Ni-NTA agarose; Qiagen) per manufacturers’ protocols. In vitro transcribed/translated Clec16a and Nrdp1 were expressed in T7-driven mammalian expression systems (Promega) per manufacturers’ protocols. Clec16a and Nrdp1 protein expression/purification was confirmed by silver staining (BioRad) and/or Western blotting. Ubiquitination assays- Detection of protein ubiquitination in
Recommended publications
  • USP8 Antibody A
    Revision 1 C 0 2 - t USP8 Antibody a e r o t S Orders: 877-616-CELL (2355) [email protected] Support: 877-678-TECH (8324) 8 2 Web: [email protected] 7 www.cellsignal.com 8 # 3 Trask Lane Danvers Massachusetts 01923 USA For Research Use Only. Not For Use In Diagnostic Procedures. Applications: Reactivity: Sensitivity: MW (kDa): Source: UniProt ID: Entrez-Gene Id: WB, IP H Mk Endogenous 130 Rabbit P40818 9101 Product Usage Information 6. Mizuno, E. et al. (2005) Mol Biol Cell 16, 5163-74. 7. Mizuno, E. et al. (2007) Exp Cell Res 313, 3624-34. Application Dilution 8. Row, P.E. et al. (2006) J Biol Chem 281, 12618-24. 9. Berlin, I. et al. (2010) J Biol Chem 285, 34909-21. Western Blotting 1:1000 Immunoprecipitation 1:50 Storage Supplied in 10 mM sodium HEPES (pH 7.5), 150 mM NaCl, 100 µg/ml BSA and 50% glycerol. Store at –20°C. Do not aliquot the antibody. Specificity / Sensitivity USP8 Antibody recognizes endogenous levels of total USP8 protein. Species Reactivity: Human, Monkey Source / Purification Polyclonal antibodies are produced by immunizing animals with a synthetic peptide corresponding to residues surrounding Pro320 of human USP8 protein. Antibodies are purified by protein A and peptide affinity chromatography. Background Ubiquitinating enzymes (UBEs) catalyze protein ubiquitination, a reversible process countered by deubiquitinating enzyme (DUB) action (1,2). Five DUB subfamilies are recognized, including the USP, UCH, OTU, MJD, and JAMM enzymes. The deubiquitinating enzyme ubiquitin-specific protease 8 (USP8/UBPy) is a cysteine protease belonging to the USP/UBP subfamily.
    [Show full text]
  • Genome-Scale Single-Cell Mechanical Phenotyping Reveals Disease-Related Genes Involved in Mitotic Rounding
    ARTICLE DOI: 10.1038/s41467-017-01147-6 OPEN Genome-scale single-cell mechanical phenotyping reveals disease-related genes involved in mitotic rounding Yusuke Toyoda 1,2, Cedric J. Cattin3, Martin P. Stewart 3,4,5, Ina Poser1, Mirko Theis6, Teymuras V. Kurzchalia1, Frank Buchholz1,6, Anthony A. Hyman1 & Daniel J. Müller 3 To divide, most animal cells drastically change shape and round up against extracellular confinement. Mitotic cells facilitate this process by generating intracellular pressure, which the contractile actomyosin cortex directs into shape. Here, we introduce a genome-scale microcantilever- and RNAi-based approach to phenotype the contribution of > 1000 genes to the rounding of single mitotic cells against confinement. Our screen analyzes the rounding force, pressure and volume of mitotic cells and localizes selected proteins. We identify 49 genes relevant for mitotic rounding, a large portion of which have not previously been linked to mitosis or cell mechanics. Among these, depleting the endoplasmic reticulum-localized protein FAM134A impairs mitotic progression by affecting metaphase plate alignment and pressure generation by delocalizing cortical myosin II. Furthermore, silencing the DJ-1 gene uncovers a link between mitochondria-associated Parkinson’s disease and mitotic pressure. We conclude that mechanical phenotyping is a powerful approach to study the mechanisms governing cell shape. 1 Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany. 2 Division of Cell Biology, Life Science Institute, Kurume University, Hyakunen-Kohen 1-1, Kurume, Fukuoka 839-0864, Japan. 3 Department of Biosystems Science and Engineering (D-BSSE), Eidgenössische Technische Hochschule (ETH) Zurich, Mattenstrasse 26, 4058 Basel, Switzerland.
    [Show full text]
  • Hereditary Spastic Paraplegia: from Genes, Cells and Networks to Novel Pathways for Drug Discovery
    brain sciences Review Hereditary Spastic Paraplegia: From Genes, Cells and Networks to Novel Pathways for Drug Discovery Alan Mackay-Sim Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia; a.mackay-sim@griffith.edu.au Abstract: Hereditary spastic paraplegia (HSP) is a diverse group of Mendelian genetic disorders affect- ing the upper motor neurons, specifically degeneration of their distal axons in the corticospinal tract. Currently, there are 80 genes or genomic loci (genomic regions for which the causative gene has not been identified) associated with HSP diagnosis. HSP is therefore genetically very heterogeneous. Finding treatments for the HSPs is a daunting task: a rare disease made rarer by so many causative genes and many potential mutations in those genes in individual patients. Personalized medicine through genetic correction may be possible, but impractical as a generalized treatment strategy. The ideal treatments would be small molecules that are effective for people with different causative mutations. This requires identification of disease-associated cell dysfunctions shared across geno- types despite the large number of HSP genes that suggest a wide diversity of molecular and cellular mechanisms. This review highlights the shared dysfunctional phenotypes in patient-derived cells from patients with different causative mutations and uses bioinformatic analyses of the HSP genes to identify novel cell functions as potential targets for future drug treatments for multiple genotypes. Keywords: neurodegeneration; motor neuron disease; spastic paraplegia; endoplasmic reticulum; Citation: Mackay-Sim, A. Hereditary protein-protein interaction network Spastic Paraplegia: From Genes, Cells and Networks to Novel Pathways for Drug Discovery. Brain Sci. 2021, 11, 403.
    [Show full text]
  • Genome-Wide DNA Methylation Dynamics During Epigenetic
    Gómez‑Redondo et al. Clin Epigenet (2021) 13:27 https://doi.org/10.1186/s13148‑021‑01003‑x RESEARCH Open Access Genome‑wide DNA methylation dynamics during epigenetic reprogramming in the porcine germline Isabel Gómez‑Redondo1*† , Benjamín Planells1†, Sebastián Cánovas2,3, Elena Ivanova4, Gavin Kelsey4,5 and Alfonso Gutiérrez‑Adán1 Abstract Background: Prior work in mice has shown that some retrotransposed elements remain substantially methylated during DNA methylation reprogramming of germ cells. In the pig, however, information about this process is scarce. The present study was designed to examine the methylation profles of porcine germ cells during the time course of epigenetic reprogramming. Results: Sows were artifcially inseminated, and their fetuses were collected 28, 32, 36, 39, and 42 days later. At each time point, genital ridges were dissected from the mesonephros and germ cells were isolated through magnetic‑ activated cell sorting using an anti‑SSEA‑1 antibody, and recovered germ cells were subjected to whole‑genome bisulphite sequencing. Methylation levels were quantifed using SeqMonk software by performing an unbiased analysis, and persistently methylated regions (PMRs) in each sex were determined to extract those regions showing 50% or more methylation. Most genomic elements underwent a dramatic loss of methylation from day 28 to day 36, when the lowest levels were shown. By day 42, there was evidence for the initiation of genomic re‑methylation. We identifed a total of 1456 and 1122 PMRs in male and female germ cells, respectively, and large numbers of transpos‑ able elements (SINEs, LINEs, and LTRs) were found to be located within these PMRs. Twenty‑one percent of the introns located in these PMRs were found to be the frst introns of a gene, suggesting their regulatory role in the expression of these genes.
    [Show full text]
  • Differential Microrna Expression in USP8-Mutated and Wild-Type Corticotroph Pituitary Tumors Reflect the Difference in Protein Ubiquitination Processes
    Journal of Clinical Medicine Article Differential microRNA Expression in USP8-Mutated and Wild-Type Corticotroph Pituitary Tumors Reflect the Difference in Protein Ubiquitination Processes Mateusz Bujko 1 , Paulina Kober 1, Joanna Boresowicz 1, Natalia Rusetska 1, Natalia Zeber-Lubecka 2, Agnieszka Paziewska 3, Monika Pekul 4, Grzegorz Zielinski 5, Andrzej Styk 5, Jacek Kunicki 6, Jerzy Ostrowski 2,7, Janusz A Siedlecki 1 and Maria Maksymowicz 4,* 1 Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; [email protected] (M.B.); [email protected] (P.K.); [email protected] (J.B.); [email protected] (N.R.); [email protected] (J.A.S.) 2 Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Centre for Postgraduate Education, 01-813 Warsaw, Poland; [email protected] (N.Z.-L.); [email protected] (J.O.) 3 Department of Neuroendocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland; [email protected] 4 Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Center, 02-781 Warsaw, Poland; [email protected] 5 Department of Neurosurgery, Military Institute of Medicine, 04-141 Warsaw, Poland; [email protected] (G.Z.); [email protected] (A.S.) 6 Department of Neurosurgery, Maria Sklodowska-Curie Institute-Oncology Center, 02-781 Warsaw, Poland; [email protected] Citation: Bujko, M.; Kober, P.; 7 Department of Genetics, Maria Sklodowska-Curie Institute-Oncology Center, 02-781 Warsaw, Poland Boresowicz, J.; Rusetska, N.; * Correspondence: [email protected] or [email protected] Zeber-Lubecka, N.; Paziewska, A.; Pekul, M.; Zielinski, G.; Styk, A.; Abstract: Background: USP8 mutations are the most common driver changes in corticotroph pituitary Kunicki, J.; et al.
    [Show full text]
  • Identification of a Novel RASD1 Somatic Mutation in a USP8-Mutated Corticotroph Adenoma
    Downloaded from molecularcasestudies.cshlp.org on September 27, 2021 - Published by Cold Spring Harbor Laboratory Press TITLE PAGE Title: Identification of a novel RASD1 somatic mutation in a USP8-mutated corticotroph adenoma. Running title: Novel RASD1 mutation in a corticotroph adenoma. Article Type: Research Report Authors and affiliations: Andrew V. Uzilov 1 Khadeen C. Cheesman* 2 Marc Y. Fink* 1 Leah C. Newman 1 Chetanya Pandya 1 Yelena Lalazar 2 Marco Hefti 3 Mary Fowkes 3 Gintaras Deikus 1 Chun Yee Lau 1 Aye S. Moe 1 Yayoi Kinoshita 3 Yumi Kasai 1 4 Micol Zweig 1 Arpeta Gupta 2 Daniela Starcevic 1 Milind Mahajan 1 Eric E. Schadt 1 Kalmon D. Post 5 Michael J. Donovan 3 Robert Sebra 1 Rong Chen# 1 Eliza B. Geer# 2,5,6 * Contributed equally to this work. # corresponding author, [email protected], [email protected] 1) Department of Genetics and Genomic Sciences and Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA 2) Division of Endocrinology, Diabetes, and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA Downloaded from molecularcasestudies.cshlp.org on September 27, 2021 - Published by Cold Spring Harbor Laboratory Press 3) Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA 4) Current affiliation: New York Genome Center, New York, NY 10013, USA 5) Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA 6) Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan Kettering, 1275 York Avenue, New York, NY 10065, USA ABSTRACT Cushing’s Disease is caused by pituitary corticotroph adenomas that secrete excess adrenocorticotropic hormone.
    [Show full text]
  • Update on the Genetics of Spastic Paraplegias Maxime Boutry, Sara Morais, Giovanni Stevanin
    Update on the genetics of spastic paraplegias Maxime Boutry, Sara Morais, Giovanni Stevanin To cite this version: Maxime Boutry, Sara Morais, Giovanni Stevanin. Update on the genetics of spastic paraple- gias. Current Neurology and Neuroscience Reports, Current Medicine Group, 2019, 19 (4), pp.18. 10.1007/s11910-019-0930-2. hal-02272013 HAL Id: hal-02272013 https://hal.sorbonne-universite.fr/hal-02272013 Submitted on 27 Aug 2019 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Update on the genetics of spastic paraplegias Maxime Boutry1,2*, Sara Morais1,2,3, Giovanni Stevanin1,2 1 Institut du Cerveau et de la Moelle épinière, Sorbonne Université UMR_S1127, INSERM Unit 1127, CNRS UMR7225, 75013, Paris, France 2 Ecole Pratique des Hautes Etudes (EPHE), Paris Sciences Lettres (PSL) Research University, Neurogenetics team, 75013, Paris, France 3 UnIGENe, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal *Current affiliation: Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada Corresponding author: Giovanni Stevanin [email protected] ORCID 0000-0001-9368-8657 Abstract Purpose of review Hereditary spastic paraplegias are a genetically heterogeneous group of neurological disorders.
    [Show full text]
  • POGLUT1, the Putative Effector Gene Driven by Rs2293370 in Primary
    www.nature.com/scientificreports OPEN POGLUT1, the putative efector gene driven by rs2293370 in primary biliary cholangitis susceptibility Received: 6 June 2018 Accepted: 13 November 2018 locus chromosome 3q13.33 Published: xx xx xxxx Yuki Hitomi 1, Kazuko Ueno2,3, Yosuke Kawai1, Nao Nishida4, Kaname Kojima2,3, Minae Kawashima5, Yoshihiro Aiba6, Hitomi Nakamura6, Hiroshi Kouno7, Hirotaka Kouno7, Hajime Ohta7, Kazuhiro Sugi7, Toshiki Nikami7, Tsutomu Yamashita7, Shinji Katsushima 7, Toshiki Komeda7, Keisuke Ario7, Atsushi Naganuma7, Masaaki Shimada7, Noboru Hirashima7, Kaname Yoshizawa7, Fujio Makita7, Kiyoshi Furuta7, Masahiro Kikuchi7, Noriaki Naeshiro7, Hironao Takahashi7, Yutaka Mano7, Haruhiro Yamashita7, Kouki Matsushita7, Seiji Tsunematsu7, Iwao Yabuuchi7, Hideo Nishimura7, Yusuke Shimada7, Kazuhiko Yamauchi7, Tatsuji Komatsu7, Rie Sugimoto7, Hironori Sakai7, Eiji Mita7, Masaharu Koda7, Yoko Nakamura7, Hiroshi Kamitsukasa7, Takeaki Sato7, Makoto Nakamuta7, Naohiko Masaki 7, Hajime Takikawa8, Atsushi Tanaka 8, Hiromasa Ohira9, Mikio Zeniya10, Masanori Abe11, Shuichi Kaneko12, Masao Honda12, Kuniaki Arai12, Teruko Arinaga-Hino13, Etsuko Hashimoto14, Makiko Taniai14, Takeji Umemura 15, Satoru Joshita 15, Kazuhiko Nakao16, Tatsuki Ichikawa16, Hidetaka Shibata16, Akinobu Takaki17, Satoshi Yamagiwa18, Masataka Seike19, Shotaro Sakisaka20, Yasuaki Takeyama 20, Masaru Harada21, Michio Senju21, Osamu Yokosuka22, Tatsuo Kanda 22, Yoshiyuki Ueno 23, Hirotoshi Ebinuma24, Takashi Himoto25, Kazumoto Murata4, Shinji Shimoda26, Shinya Nagaoka6, Seigo Abiru6, Atsumasa Komori6,27, Kiyoshi Migita6,27, Masahiro Ito6,27, Hiroshi Yatsuhashi6,27, Yoshihiko Maehara28, Shinji Uemoto29, Norihiro Kokudo30, Masao Nagasaki2,3,31, Katsushi Tokunaga1 & Minoru Nakamura6,7,27,32 Primary biliary cholangitis (PBC) is a chronic and cholestatic autoimmune liver disease caused by the destruction of intrahepatic small bile ducts. Our previous genome-wide association study (GWAS) identifed six susceptibility loci for PBC.
    [Show full text]
  • Systematic Elucidation of Neuron-Astrocyte Interaction in Models of Amyotrophic Lateral Sclerosis Using Multi-Modal Integrated Bioinformatics Workflow
    ARTICLE https://doi.org/10.1038/s41467-020-19177-y OPEN Systematic elucidation of neuron-astrocyte interaction in models of amyotrophic lateral sclerosis using multi-modal integrated bioinformatics workflow Vartika Mishra et al.# 1234567890():,; Cell-to-cell communications are critical determinants of pathophysiological phenotypes, but methodologies for their systematic elucidation are lacking. Herein, we propose an approach for the Systematic Elucidation and Assessment of Regulatory Cell-to-cell Interaction Net- works (SEARCHIN) to identify ligand-mediated interactions between distinct cellular com- partments. To test this approach, we selected a model of amyotrophic lateral sclerosis (ALS), in which astrocytes expressing mutant superoxide dismutase-1 (mutSOD1) kill wild-type motor neurons (MNs) by an unknown mechanism. Our integrative analysis that combines proteomics and regulatory network analysis infers the interaction between astrocyte-released amyloid precursor protein (APP) and death receptor-6 (DR6) on MNs as the top predicted ligand-receptor pair. The inferred deleterious role of APP and DR6 is confirmed in vitro in models of ALS. Moreover, the DR6 knockdown in MNs of transgenic mutSOD1 mice attenuates the ALS-like phenotype. Our results support the usefulness of integrative, systems biology approach to gain insights into complex neurobiological disease processes as in ALS and posit that the proposed methodology is not restricted to this biological context and could be used in a variety of other non-cell-autonomous communication
    [Show full text]
  • MAGEL2 Regulates Leptin Receptor Internalization Through Ubiquitination Pathways Involving USP8 and RNF41
    MAGEL2 regulates leptin receptor internalization through ubiquitination pathways involving USP8 and RNF41 by Tishani Methsala Wijesuriya A thesis submitted in partial fulfillment of the requirements for the degree of Master of Science Medical Sciences – Medical Genetics University of Alberta ©Tishani Methsala Wijesuriya, 2017 Abstract Children with Prader-Willi syndrome have neonatal feeding difficulties, developmental delay and excessive appetite. Loss of MAGEL2 alone causes a related neurodevelopmental disorder (Schaaf- Yang syndrome) and may contribute to obesity in children with Prader-Willi syndrome who lack MAGEL2 and other genes. MAGEL2 is essential in neurons that sense levels of the adipose tissue- derived hormone leptin. The MAGEL2 protein is important for recycling or degradation of proteins in the brain and interacts with and modifies the activity of E3 ubiquitin ligases. RNF41 is a E3 ubiquitin ligase that associates with a ubiquitin-specific protease (USP8). Together with USP8, RNF41 regulates the recycling of the leptin receptor by targeting it either for degradation or for recycling to the cell membrane. I hypothesized that MAGEL2 regulates the interaction between RNF41 and USP8, and that loss of this regulation could impair leptin response pathways in the brain in children with PWS. Human U2OS cells were transfected with recombinant constructs encoding epitope tagged versions of MAGEL2 and wild type and mutant forms of RNF41. Immunofluorescence was used to visualize the co-localization of different forms of RNF41 with MAGEL2 in intracellular compartments. I expressed recombinant MAGEL2, RNF41 and USP8 in combinations in human U2OS cells and examined the relative abundance of each protein in the presence or absence of the other components of the complex.
    [Show full text]
  • Full-Text.Pdf
    Systematic Evaluation of Genes and Genetic Variants Associated with Type 1 Diabetes Susceptibility This information is current as Ramesh Ram, Munish Mehta, Quang T. Nguyen, Irma of September 23, 2021. Larma, Bernhard O. Boehm, Flemming Pociot, Patrick Concannon and Grant Morahan J Immunol 2016; 196:3043-3053; Prepublished online 24 February 2016; doi: 10.4049/jimmunol.1502056 Downloaded from http://www.jimmunol.org/content/196/7/3043 Supplementary http://www.jimmunol.org/content/suppl/2016/02/19/jimmunol.150205 Material 6.DCSupplemental http://www.jimmunol.org/ References This article cites 44 articles, 5 of which you can access for free at: http://www.jimmunol.org/content/196/7/3043.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 23, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Systematic Evaluation of Genes and Genetic Variants Associated with Type 1 Diabetes Susceptibility Ramesh Ram,*,† Munish Mehta,*,† Quang T.
    [Show full text]
  • Reciprocal Cross-Regulation Between RNF41 and USP8 Controls Cytokine
    3770 Research Article Reciprocal cross-regulation between RNF41 and USP8 controls cytokine receptor sorting and processing Leentje De Ceuninck1,2,*, Joris Wauman1,2,*, Delphine Masschaele1,2, Frank Peelman1,2 and Jan Tavernier1,2,` 1Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), Albert Baertsoenkaai 3, B-9000 Ghent, Belgium 2Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium *These authors contributed equally to this work `Author for correspondence ([email protected]) Accepted 15 May 2013 Journal of Cell Science 126, 3770–3781 ß 2013. Published by The Company of Biologists Ltd doi: 10.1242/jcs.131250 Summary The mechanisms controlling the steady-state cell surface levels of cytokine receptors, and consequently the cellular response to cytokines, remain poorly understood. The number of surface-exposed receptors is a dynamic balance of de novo synthesis, transport to the plasma membrane, internalization, recycling, degradation and ectodomain shedding. We previously reported that the E3 ubiquitin ligase RING finger protein 41 (RNF41) inhibits basal lysosomal degradation and enhances ectodomain shedding of JAK2-associated cytokine receptors. Ubiquitin-specific protease 8 (USP8), an RNF41-interacting deubiquitylating enzyme (DUB) stabilizes RNF41 and is involved in trafficking of various transmembrane proteins. The present study identifies USP8 as a substrate of RNF41 and reveals that loss of USP8 explains the aforementioned RNF41 effects. RNF41 redistributes and ubiquitylates USP8, and reduces USP8 levels. In addition, USP8 knockdown functionally matches the effects of RNF41 ectopic expression on the model leptin and leukemia inhibitory factor (LIF) receptors. Moreover, RNF41 indirectly destabilizes the ESCRT-0 complex through suppression of USP8.
    [Show full text]